Anda di halaman 1dari 7

Journal of Central Nervous System Disease

Review

Open Access
Full open access to this and
thousands of other papers at
http://www.la-press.com.

Alzheimers Disease: Review of Emerging Treatment


Role for Intravenous Immunoglobulins
Rakez Kayed13, George R.Jackson13, D. Mark Estes3,4, and Alan D.T. Barrett3,4
Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA.
Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA. 3Sealy Center for Vaccine
Development, University of Texas Medical Branch, Galveston, TX, USA. 4Department of Pathology, University
of Texas Medical Branch, Galveston, TX, USA. Corresponding author email: rakayed@utmb.edu
1
2

Abstract: Alzheimers disease (AD) is the most common neurodegenerative disorder. Currently available therapies are symptomatic
but do not alter underlying disease progression. Immunotherapeutic approaches such as anti A peptide active vaccination trials have
had limited success to date. Intravenous immunoblobulin (IVIg) is widely used in immune-mediated neurological disorders such myasthenia gravis and Guillain-Barre syndrome. These preparations have been obtained from the pooled plasma of healthy human donors
and contain natural anti-amyloid antibodies and are well tolerated. A small pilot study of passive immunotherapy using IVIg has suggested cognitive improvement. A multicenter phase III trial is ongoing and will determine whether or not this treatment can ameliorate
cognitive deficits in mild-to-moderate AD. Here, we briefly review the pathogenic role of amyloid and tau in AD, as well as immunotherapeutic efforts to date. We also summarize what is known about naturally occurring anti-A and tau antibodies in IVIg with a view
toward explaining potential mechanisms underlying their therapeutic effects.
Keywords: Alzheimer`s, immunotherapy, conformation antibodies, tau oligomers, amyloid oligomers.

Journal of Central Nervous System Disease 2011:3 6773


doi: 10.4137/JCNSD.S5018
This article is available from http://www.la-press.com.
the author(s), publisher and licensee Libertas Academica Ltd.
This is an open access article. Unrestricted non-commercial use is permitted provided the original work is properly cited.
Journal of Central Nervous System Disease 2011:3

67

Kayed etal

Introduction

Alzheimers disease (AD) is the most common


neurodegenerative disorder, with devastating personal and financial costs. In the absence of clear
prevention strategies or disease modifying therapies,
it is expected that the number of people affected
by AD worldwide will exceed 100 million in 2050.
With improved survival from acute diseases and
the increasing lifespan of populations in developed
and middle income countries, dramatic increases in
the incidence of Alzheimers disease (AD) are predicted, with dire consequences for the economic and
social fabric of many nations.1 Hence, development
of effective disease-modifying therapies for AD is
an urgent priority for research in both academia and
pharmaceutical companies. AD is a complex disease
with two principle hallmark events: 1) the misfolding,
aggregation and brain deposition of amyloid- (A)
peptide in amyloid plaques, and 2) the deposition
of misfolded tau protein in neurofibrillary tangles
(NFT).2 The A peptide is generated from the cleavage of amyloid precursor protein (APP) by and
secretases.2 Given the preeminence of the amyloid
hypothesis3 in the AD field, extensive efforts have
targeted various forms of A aggregates for drug
development; these include reduction and alteration
of APP processing, prevention of A misfolding
and aggregation, minimization or elimination of
its neurotoxicity, acceleration of its clearance and
degradation,49 as well as active (ie, stimulation of
an immune response following administration of an
immunogen), and passive (ie, provision of short term
protection against infection or clinical condition by
administration of antibodies) vaccination strategies to
remove amyloid deposits.10
An ever-increasing body of evidence implicating
tau in neurodegenerative diseases11,12 supports tau
as a potential target for the development of diseasemodifying therapeutics.13,14 Tau-based therapeutic
approaches have historically lagged behind anti A
approaches. Recently, however, tau-based approaches
have been the subject of renewed interest;13 potential therapeutics may manipulate tau via inhibition of
phosphorylation,15,16 activation of proteolytic or proteasomal degradation pathways,17,18 microtubule-binding
drugs (eg, paclitaxel) for stabilization of microtubule networks,19,20 inhibition of aggregation by small
molecules,21,22 or clearance by immunotherapy.2326
68

Immunotherapy Against A

Arguably the most exciting treatment approach


for AD to have evolved recently is anti-A
immunotherapy using antibodies to A administered on multiple occasions.27 First introduced
by Schenk and colleagues in 1999, promising
results were described initially in animal models.28
Ten years later, enthusiasm for anti A immunotherapy has been largely replaced by frustration due to
adverse effects including meningoencephalitis and
leukoencephalopathy.10,2931 Results from the ongoing phase II trial of anti A humanized monoclonal
antibody (Bapineuzumab) have confirmed removal
of amyloid plaques as detected by positron emission
tomography (PET) scans using Pittsburgh compound
B (11C-PiB), but without concomitant cognition
improvement.32 In addition, despite evidence of amyloid plaque removal,33 post mortem analysis of brains
from those patients has failed to demonstrate changes
in tau pathology, neuropil threads, synaptic dysfunction, or cerebral amyloid angiopathy.29,34,35 Despite
these disappointments, the research community has
persevered with alternative regimens of administration in efforts to develop and optimize a more effective passive or active A-based vaccine. These efforts
have included the use of IVIg preparations that contain naturally occurring anti A antibodies.3638
Recent literature reveals a shift in focus from cure
toward understanding mechanisms associated with
benefits in animal models and etiology of complications reported in both humans and animal models
(reviewed in).3944

Conformation-Specific Antibodies

Amyloid diseases, including many neurodegenerative


disorders, are considered conformational diseases,
since amyloid formation is triggered by conformational
changes in a specific peptide or protein, resulting
in its misfolding and deposition as amyloid.4547
Moreover, conformation-specific antibodies that
recognize specific amyloid species, eg, fibrils or oligomers, from many types of amyloid proteins have
been produced and characterized.4850 Conformationspecific antibodies were derived from observations
reported more than thirty years ago51,52 indicating that
amyloid antibodies react with conformational epitopes
and not with native protein structure, ie, suggesting
that amyloid fibrils have a non-native structure.51,52
Journal of Central Nervous System Disease 2011:3

Immunoglobulin therapies for Alzheimers disease

Numerous conformation-specific antibodies have


been generated and characterized, including a few
that are commercially available. Such antibodies
have been used to characterize disease progression
and to ameliorate amyloid toxicity (see review by
Glabe).50 Moreover, conformation-specific antibody
domains and single chain fragment variable (scFv)
constructs with similar specificity have been reported;
of note, these can cross the blood-brain barrier more
efficiently than antibodies and can be expressed
intracellularly.5355
The critical role of soluble amyloid oligomers
in neurodegeneration has become more generally
accepted for multiple neurodegenerative diseases,
including AD.5660 Results obtained using oligomeric
conformation-specific antibodies49 indicate that
oligomers (protofibrils) have a common, generic structure that is distinct from both fibrils and low molecular
weight soluble monomer/dimers. Furthermore, such
antibodies recognize soluble oligomers from a variety of different amyloids, including lysozyme, islet
amyloid polypeptide (IAPP), synuclein, prion protein, polyglutamine, and insulin. The anti oligomer
antibody (A11) that binds specifically to amyloid
oligomers49 has more robust effects as compared to
other anti amyloid antibodies when injected intrathecally into the TgCRND8 AD mouse model.61 Surprisingly, similar conformation-specific antibodies have
been detected in humans using peptide microarrays.
Britschgi etal demonstrated the presence of sequenceindependent, oligomeric conformational antibodies
in human plasma and CSF.62 Although the diversity,
abundance, and function of such endogenous conformational antibodies remain largely uncharacterized,
these investigators have reported that these antibodies decline with age and advancing AD, suggesting
that they may play a role in protection against toxic
amyloid oligomers.62

Tau-Based Immunotherapy

Tau immunotherapy is a new concept.23 To date, only


three reports of tau immunotherapy in animal models
have been published, all using active vaccination.24,25,63
To date, no reports of passive vaccination have
appeared. In the first report, the authors used a tau
fragment (379408) phosphorylated at Ser396 and
Ser404 (phosphorylation sites commonly associated
with NFT) to vaccinate the P301L mouse model.64
Journal of Central Nervous System Disease 2011:3

Behavioral analysis showed improved performance


after immunization as compared to controls. These
data demonstrated that antibodies against this immunogen were able to cross the blood-brain barrier
and bind to phosphorylated tau.24 The Rosenmann
group used phosphorylated tau with Freunds adjuvant and pertussis toxin adjuvants; these investigators reported a 40% reduction in NFTs and 20%
increase in microglia.25 In 2006, the Rosenmann
group also reported that full-length tau was encephalitogenic, triggering a severe autoimmune response.63
Mice vaccinated with soluble tau developed NFTlike structures, axonal damage, gliosis, mononuclear
infiltrates, and motor phenotypes. These data demonstrate the potential dangers of using soluble tau as
immunogen, or of antibodies recognizing epitopes of
full-length tau for passive vaccination. Although the
use of phosphorylated tau antigens seems promising
for vaccination studies (ie, presenting specific phosphoepitopes to the immune system), such an approach
has significant potential risks, as these phosphorylation sites are mainly associated with NFT.65,66 An
optimal vaccine should target pre-filament tau species
(tau oligomers), which form at early stages of NFT
development rather than mature, meta-stable NFT.26
Pre-filament specific phosphorylation sites have yet
to be conclusively identified due to the complexity
of tau aggregation, the overlap between the three
stages of NFTs development with regard to tau phosphorylation sites,65 and the fact that tau phosphorylation is a physiological process that is essential for tau
normal function and reversible.67 In vitro assembled
tau paired helical filaments (PHF) similar to those
found in AD have -structure similar to other amyloid
fibrils.68,69 Moreover, tau conformation-specific antibodies (eg, Alz50 and MC-1) recognize conformational tau epitopes associated with PHF.70,71 Naturally
occurring antibodies have been detected in the blood
of normal and AD patients, including antibodies
against both unphosphorylated and phosphorylated
tau.72 Both IgG and IgM anti-tau antibodies have
been identified in serum collected from AD patients
and controls; very few tau antibodies are found in
CSF. Higher anti-phosphorylated-tau IgM antibodies
have been found in AD patients relative to controls.
In this work of Steinitz and coworkers, all subjects
(both AD and controls) were greater than 70 years
of age;72 it is reasonable to assume that higher levels
69

Kayed etal

of anti-phosphotau IgM would be present in healthy


young controls, as reported for A antibodies. If so,
it seems likely that larger studies will confirm these
findings and identify higher concentrations of neuroprotective tau antibodies in young healthy people.

IVIg Immunotherapy

Intravenous immunoglobulin (IVIg) is an antibody


product obtained from human plasma from thousands
of donors. For more than thirty years, IVIg has been
used for the treatment of post-exposure to infectious
diseases, immune disorders and the management
of patients with neurological conditions.7376 IVIg
treatment is used routinely for some immune-mediated
neurological disorders such as Guillain-Barre syndrome, patients treated with IVIg have reduced risk
of developing Alzheimers disease and recently IVIg
has been investigated for the treatment of neurodegenerative disorders.77 Commercially available IVIg
has been used in small pilot trial in AD.36 In this open
label study, 8mildly affected patients received IVIg
for a total of 15months over two intervals (6months,
discontinued for 3 months, then followed by
9months further treatment). Infusions were generally
well-tolerated; of note, both anti-A antibodies and
A levels in the serum increased after each infusion
in proportion to IVIg dose, whereas A levels in CSF
decreased significantly at 6months, returned to baseline after washout, and decreased again after IVIg
was re-administered for the additional 9 months.
Mini-mental state scores increased an average of
2.5 points after 6months, returned to baseline during
washout, and remained stable during subsequent
9 months treatment.36 A smaller study previously
had shown positive effects in five AD patients, each
of whom received one dose of IVIg per month for
6months.38 Taken together, these findings justified a
large-scale randomized phase III clinical trial that has
enrolled more than 360AD patients.76,78
Naturally occurring antibodies that can detect
and block the toxicity of special conformations displayed by misfolded proteins, including amyloid-,
-synuclein, and prion protein, are detectable in
human serum and CSF. Although such antibodies
are detectable in both healthy individuals and
patients, it is clear that their levels are reduced
in the latter.62,7882 However, an assay for precise
quantification of endogenous human A antibodies
70

is not yet available. Efforts are underway to identify


and overcome factors contributing to wide disparities
among reported measurements.8385 Such efforts seem
likely to improve the quality of IVIg preparations for
AD treatment, and they may lead to the development
of methods to isolate specific anti-amyloid antibody
populations that can then be tested for their potential
to treat AD and other neurodegenerative diseases by
passive administration.80,8385 Humanized monoclonal
antibodies have applications but they recognize only a
single epitope whereas naturally occurring antibodies
are polyclonal and will recognize multiple epitopes,
and more likely to have stronger therapeutic effects.

Which Antibodies Should We be


Looking for in IVIg Preparations?

In addition to the anti A oligomer antibodies


reported in IVIg,82,83 these preparations may contain
neuroprotective anti tau oligomer antibodies that
may account, at least in part, for the positive results
derived from IVIg treatment in AD. Recently, tau
oligomers have emerged as a likely pathogenic entity
in tauopathies. Although their formation and role in
neurodegeneration has yet to be fully elucidated, an
increasing literature argues that they play a crucial
role in AD and other tauopathies. Stereological
analysis of AD demonstrates that neuronal loss
actually precedes NFT formation.86,87 This observation, as well as data emerging from biochemical,
cell-based and transgenic mouse studies, suggests
that soluble tau aggregates may be the most toxic
and pathologically significant tau species.57,8892
Studies using different animal models suggest that
tau oligomers play a key role in impaired synaptic function, hippocampal synapse loss, and microgliosis leading to neurodegeneration and behavioral
impairments. All of these phenomena are concurrent with accumulation of soluble aggregated tau
species and dissociated from the accumulation
of NFT.57,93,94 Tau oligomers have been characterized biochemically in a conditional mouse model
(rTg4510) expressing the P301L human tau mutant;
surprisingly, the accumulation of oligomeric tau
(but not NFT) correlated with neuronal loss and
behavioral deficits in this model.95,96 Tau oligomers
have been characterized in human brain; a correlation between disease progression and the accumulation of granular tau oligomers in the brains of AD
Journal of Central Nervous System Disease 2011:3

Immunoglobulin therapies for Alzheimers disease

patients has been reported.97,98 Moreover, increased


levels of tau oligomers are detected in the frontal
cortex at very early stage of the disease (Braak
stage I), before clinical manifestations of AD and
NFT are believed to develop.97,98 Given the heterogeneity of tau species, ranging from monomer to
oligomer, to PHF/NFT, it seems prudent that future
efforts should focus on complete characterization
of all tau antibodies present in CSF, rather than on
any particular species. Meticulous analysis of both
anti tau and anti amyloid antibodies present in IVIg
preparations is critical for a better understanding of
mechanisms underlying potential benefits of such
preparations in AD.

Conclusions

The results from the pilot clinical trial justify further


investigation of IVIg for the treatment of AD.
Further examination of endogenous neuroprotective
antibodies should help us to better understand their
mechanism of action. Given that anti-A immunotherapy trials were disappointing, it seems unlikely
that naturally occurring A antibodies are likely to
account for observed clinical benefits of IVIg; further,
such IVIg preparations may be effective because they
contain both anti-tau and anti-A natural antibodies.
Apart from the presence of these antibodies, IVIg may
be useful for AD treatment due to anti-inflammatory
effects at high doses mediated in part via Fc receptor
uptake of IgG by inflammatory cells.73,74 IVIg is
a very expensive treatment, and despite its safety
record in the treatment of multiple neurological
disorders, potential complications include headache,
dermatitis, infection, pulmonary edema, allergic/
anaphylactic reactions, acute renal failure, venous
thrombosis, and aseptic meningitis.99,100 These side
effects may hinder the use of IVIg in AD, which has
a long asymptomatic phase and survival period once
symptomatic. Therefore, critical analysis of IVIg
preparations and further examination of endogenous
neuroprotective antibodies are necessary to better understand their mechanism of action and optimize their role as a disease modifying therapeutic in
AD and other neurodegenerative diseases. Finally,
significant advances have been made in the use of
humanized monoclonal antibodies for passive vaccination against infectious diseases that will do doubt
have applications.
Journal of Central Nervous System Disease 2011:3

Disclosure

This manuscript has been read and approved by


all authors. This paper is unique and is not under
consideration by any other publication and has not
been published elsewhere. The authors and peer
reviewers of this paper report no conflicts of interest.
The authors confirm that they have permission to
reproduce any copyrighted material.

References

1. Editorial. How much is dementia care worth? The Lancet Neurology. 2010;
9:1037.
2. Selkoe DJ. Alzheimers disease: Genes, proteins, and therapy. Physiol Rev.
2001;81:74166.
3. Hardy J, Selkoe DJ. The amyloid hypothesis of Alzheimers disease: progress
and problems on the road to therapeutics. Science. 2002;297:3536.
4. Roberson ED, Mucke L. 100 years and counting: prospects for defeating
Alzheimers disease. Science. 2006;314:7814.
5. Shah RS, Lee HG, Xiongwei Z, Perry G, Smith MA, Castellani RJ. Current
approaches in the treatment of Alzheimers disease. Biomed Pharmacother.
2008;62:199207.
6. Stains CI, Mondal K, Ghosh I. Molecules that target beta-amyloid. Chem
Med Chem. 2007;2:167492.
7. Klaver DW, Wilce MC, Cui H, Hung AC, Gasperini R, Foa L, etal. Is BACE1
a suitable therapeutic target for the treatment of Alzheimers disease? Current
strategies and future directions. Biol Chem. 2010;391:84959.
8. Kovacs T. Therapy of Alzheimer disease. Neuropsychopharmacol Hung.
2009;11:2733.
9. Wolfe MS. Inhibition and modulation of gamma-secretase for Alzheimers
disease. Neurotherapeutics. 2008;5:3918.
10. Wisniewski T, Boutajangout A. Vaccination as a therapeutic approach to
alzheimers disease. Mt Sinai J Med. 2010;77:1731.
11. Ballatore C, Lee VM, Trojanowski JQ. Tau-mediated neurodegeneration
in Alzheimers disease and related disorders. Nat Rev Neurosci. 2007;8:
66372.
12. Haroutunian V, Davies P, Vianna C, Buxbaum JD, Purohit DP. Tau protein
abnormalities associated with the progression of alzheimer disease type
dementia. Neurobiol Aging. 2007;28:17.
13. Schneider A, Mandelkow E. Tau-based treatment strategies in neurodegenerative diseases. Neurotherapeutics. 2008;5:44357.
14. Iqbal K, Liu F, Gong CX, Alonso AD, Grundke-Iqbal I. Mechanisms of
tau-induced neurodegeneration. Acta Neuropathol. 2009;118:5369.
15. Iqbal K, Grundke-Iqbal I. Tau phosphatase activity as a therapeutic target
for AD. Drug News Perspect. 1998;11:104.
16. Iqbal K, Grundke-Iqbal I. Inhibition of neurofibrillary degeneration:
a promising approach to Alzheimers disease and other tauopathies.
Curr Drug Targets. 2004;5:495502.
17. Johnson GV, Jope RS, Binder LI. Proteolysis of tau by calpain. Biochem
Biophys Res Commun. 1989;163:150511.
18. Sengupta S, Horowitz PM, Karsten SL, Jackson GR, Geschwind DH, Fu Y,
etal. Degradation of tau protein by puromycin-sensitive aminopeptidase in
vitro. Biochemistry. 2006;45:151119.
19. Trojanowski JQ, Smith AB, Huryn D, Lee VM. Microtubule-stabilising
drugs for therapy of Alzheimers disease and other neurodegenerative
disorders with axonal transport impairments. Expert Opin Pharmacother.
2005;6:6836.
20. Zhang B, Maiti A, Shively S, Lakhani F, McDonald-Jones G, Bruce J,
et al. Microtubule-binding drugs offset tau sequestration by stabilizing
microtubules and reversing fast axonal transport deficits in a tauopathy
model. Proc Natl Acad Sci U S A. 2005;102:22731.
21. Bulic B, Pickhardt M, Schmidt B, Mandelkow EM, Waldmann H,
Mandelkow E. Development of tau aggregation inhibitors for Alzheimers
disease. Angew Chem Int Ed Engl. 2009;48:174052.

71

Kayed etal
22. Duff K, Kuret J, Congdon EE. Disaggregation of Tau as a Therapeutic
Approach to Tauopathies. Curr Alzheimer Res. 2010;7:23540.
23. Sigurdsson EM. Tau-focused immunotherapy for Alzheimers disease and
related tauopathies. Curr Alzheimer Res. 2009;6:44650.
24. Asuni AA, Boutajangout A, Quartermain D, Sigurdsson EM. Immuno
therapy targeting pathological tau conformers in a tangle mouse model
reduces brain pathology with associated functional improvements.
J Neurosci. 2007;27:911529.
25. Boimel M, Grigoriadis N, Lourbopoulos A, Haber E, Abramsky O,
Rosenmann H. Efficacy and safety of immunization with phosphorylated
tau against neurofibrillary tangles in mice. Exp Neurol. 2010;224:47285.
26. Kayed R, Jackson GR. Prefilament tau species as potential targets for immunotherapy for Alzheimer disease and related disorders. Curr Opin Immunol.
2009;21:35963.
27. Wisniewski T, Boutajangout A. Immunotherapeutic approaches for
Alzheimers disease in transgenic mouse models. Brain Struct Funct. 2010;
214:20118.
28. Schenk D, Barbour R, Dunn W, Gordon G, Grajeda H, Guido T, etal. Immunization with amyloid-beta attenuates Alzheimer-disease-like pathology in
the PDAPP mouse. Nature. 1999;400:1737.
29. Holmes C, Boche D, Wilkinson D, Yadegarfar G, Hopkins V, Bayer A,
etal. Long-term effects of Abeta42 immunisation in Alzheimers disease:
follow-up of a randomised, placebo-controlled phase I trial. Lancet. 2008;
372:21623.
30. Nicoll JA, Barton E, Boche D, Neal JW, Ferrer I, Thompson P, etal. Abeta
species removal after abeta42 immunization. J Neuropathol Exp Neurol.
2006;65:10408.
31. Weiner HL, Frenkel D. Immunology and immunotherapy of Alzheimers
disease. Nat Rev Immunol. 2006;6:40416.
32. Rinne JO, Brooks DJ, Rossor MN, Fox NC, Bullock R, Klunk WE, etal.
11C-PiB PET assessment of change in fibrillar amyloid-beta load in patients
with Alzheimers disease treated with bapineuzumab: a phase 2, doubleblind, placebo-controlled, ascending-dose study. Lancet Neurol. 2010;9:
36372.
33. Schenk D. Amyloid-beta immunotherapy for Alzheimers disease: the end
of the beginning. Nat Rev Neurosci. 2002;3:8248.
34. Nicoll JA, Wilkinson D, Holmes C, Steart P, Markham H, Weller RO.
Neuropathology of human Alzheimer disease after immunization with
amyloid-beta peptide: a case report. Nat Med. 2003;9:44852.
35. Boche D, Zotova E, Weller RO, Love S, Neal JW, Pickering RM, et al.
Consequence of Abeta immunization on the vasculature of human
Alzheimers disease brain. Brain. 2008;131:3299310.
36. Relkin NR, Szabo P, Adamiak B, Burgut T, Monthe C, Lent RW, et al.
18-Month study of intravenous immunoglobulin for treatment of mild
Alzheimer disease. Neurobiol Aging. 2009;30:172836.
37. Weksler ME, Pawelec G, Franceschi C. Immune therapy for age-related
diseases. Trends Immunol. 2009;30:34450.
38. Dodel RC, Du Y, Depboylu C, Hampel H, Frolich L, Haag A, et al.
Intravenous immunoglobulins containing antibodies against beta-amyloid
for the treatment of Alzheimers disease. J Neurol Neurosurg Psychiatry.
2004;75:14724.
39. Woodhouse A, Dickson TC, Vickers JC. Vaccination strategies for
Alzheimers disease: A new hope? Drugs Aging. 2007;24:10719.
40. Wilcock DM, Colton CA. Anti-amyloid-beta immunotherapy in Alzheimers
disease: relevance of transgenic mouse studies to clinical trials. J Alzheimers
Dis. 2008;15:55569.
41. Brendza RP, Holtzman DM. Amyloid-beta immunotherapies in mice and
men. Alzheimer Dis Assoc Disord. 2006;20:11823.
42. Brody DL, Holtzman DM. Active and passive immunotherapy for neurodegenerative disorders. Annu Rev Neurosci. 2008;31:17593.
43. Lichtlen P, Mohajeri MH. Antibody-based approaches in Alzheimers
research: safety, pharmacokinetics, metabolism, and analytical tools.
J Neurochem. 2008;104:85974.
44. Schenk DB, Seubert P, Grundman M, Black R. A beta immunotherapy:
Lessons learned for potential treatment of Alzheimers disease. Neurodegener
Dis. 2005;2:25560.

72

45. Zerovnik E. Protein conformational pathology in Alzheimers and other


neurodegenerative diseases; new targets for therapy. Curr Alzheimer Res.
2010;7:7483.
46. Reiss C, Ehrlich R, Lesnick T, Parvez S, Parvez H. Conformational diseases:
misfolding mechanisms may pave the way to early therapy. Neurotoxicol
Teratol. 2002;24:ixxiv.
47. Chiti F, Dobson CM. Protein misfolding, functional amyloid, and human
disease. Annu Rev Biochem. 2006;75:33366.
48. Kayed R, Head E, Sarsoza F, Saing T, Cotman CW, Necula M, etal. Fibril
specific, conformation dependent antibodies recognize a generic epitope
common to amyloid fibrils and fibrillar oligomers that is absent in prefibrillar
oligomers. Mol Neurodegener. 2007;2:18.
49. Kayed R, Head E, Thompson JL, McIntire TM, Milton SC, Cotman CW,
et al. Common structure of soluble amyloid oligomers implies common
mechanism of pathogenesis. Science. 2003;300:4869.
50. Glabe CG. Conformation-dependent antibodies target diseases of protein
misfolding. Trends Biochem Sci. 2004;29:5427.
51. Franklin EC, Zucker-Franklin D. Antisera specific for human amyloid
reactive with conformational antigens. Proc Soc Exp Biol Med. 1972;140:
5658.
52. Linke RP, Zucker-Franklin D, Franklin ED. Morphologic, chemical, and
immunologic studies of amyloid-like fibrils formed from Bence Jones
Proteins by proteolysis. J Immunol. 1973;111:1023.
53. Robert R, Dolezal O, Waddington L, Hattarki MK, Cappai R, Masters CL,
et al. Engineered antibody intervention strategies for Alzheimers disease
and related dementias by targeting amyloid and toxic oligomers. Protein
Eng Des Sel. 2009;22:199208.
54. Habicht G, Haupt C, Friedrich RP, Hortschansky P, Sachse C, Meinhardt J,
etal. Directed selection of a conformational antibody domain that prevents
mature amyloid fibril formation by stabilizing Abeta protofibrils. Proc Natl
Acad Sci U S A. 2007;104:192327.
55. Barkhordarian H, Emadi S, Schulz P, Sierks MR. Isolating recombinant antibodies against specific protein morphologies using atomic force microscopy
and phage display technologies. Protein Eng Des Sel. 2006;19:497502.
56. Haass C, Selkoe DJ. Soluble protein oligomers in neurodegeneration:
lessons from the Alzheimers amyloid beta-peptide. Nat Rev Mol Cell Biol.
2007;8:10112.
57. Brunden KR, Trojanowski JQ, Lee VM. Evidence that non-fibrillar tau
causes pathology linked to neurodegeneration and behavioral impairments.
J Alzheimers Dis. 2008;14:3939.
58. Caughey B, Baron GS, Chesebro B, Jeffrey M. Getting a grip on prions:
oligomers, amyloids, and pathological membrane interactions. Annu Rev
Biochem. 2009;78:177204.
59. Glabe CG. Common mechanisms of amyloid oligomer pathogenesis in
degenerative disease. Neurobiol Aging. 2006;27:5705.
60. Glabe CG. Structural classification of toxic amyloid oligomers. J Biol
Chem. 2008;283:2963943.
61. Chauhan NB. Intracerebroventricular passive immunization with
anti-oligoAbeta antibody in TgCRND8. J Neurosci Res. 2007;85:45163.
62. Britschgi M, Olin CE, Johns HT, Takeda-Uchimura Y, LeMieux MC,
Rufibach K, etal. Neuroprotective natural antibodies to assemblies of amyloidogenic peptides decrease with normal aging and advancing Alzheimers
disease. Proc Natl Acad Sci U S A. 2009;106:1214550.
63. Rosenmann H, Grigoriadis N, Karussis D, Boimel M, Touloumi O,
Ovadia H, etal. Tauopathy-like abnormalities and neurologic deficits in mice
immunized with neuronal tau protein. Arch Neurol. 2006;63:145967.
64. Lewis J, McGowan E, Rockwood J, Melrose H, Nacharaju P,
van Slegtenhorst M, et al. Neurofibrillary tangles, amyotrophy and progressive motor disturbance in mice expressing mutant (P301L) tau protein.
Nat Genet. 2000;25:4025.
65. Augustinack JC, Schneider A, Mandelkow EM, Hyman BT. Specific tau
phosphorylation sites correlate with severity of neuronal cytopathology in
Alzheimers disease. Acta Neuropathol. 2002;103:2635.
66. Trinczek B, Biernat J, Baumann K, Mandelkow EM, Mandelkow E.
Domains of tau protein, differential phosphorylation, and dynamic instability of microtubules. Mol Biol Cell. 1995;6:1887902.

Journal of Central Nervous System Disease 2011:3

Immunoglobulin therapies for Alzheimers disease


67. Su B, Wang X, Drew KL, Perry G, Smith MA, Zhu X. Physiological
regulation of tau phosphorylation during hibernation. J Neurochem. 2008;
105:2098108.
68. Berriman J, Serpell LC, Oberg KA, Fink AL, Goedert M, Crowther RA. Tau
filaments from human brain and from in vitro assembly of recombinant protein show cross-beta structure. Proc Natl Acad Sci U S A. 2003;100:90348.
69. Barghorn S, Davies P, Mandelkow E. Tau paired helical filaments from
Alzheimers disease brain and assembled in vitro are based on beta-structure
in the core domain. Biochemistry. 2004;43:1694703.
70. Weaver CL, Espinoza M, Kress Y, Davies P. Conformational change as one
of the earliest alterations of tau in Alzheimers disease. Neurobiol Aging.
2000;21:71927.
71. Jicha GA, Bowser R, Kazam IG, Davies P. Alz-50 and MC-1, a new
monoclonal antibody raised to paired helical filaments, recognize conformational epitopes on recombinant tau. J Neurosci Res. 1997;48:12832.
72. Rosenmann H, Meiner Z, Geylis V, Abramsky O, Steinitz M. Detection of
circulating antibodies against tau protein in its unphosphorylated and in its
neurofibrillary tangles-related phosphorylated state in Alzheimers disease
and healthy subjects. Neurosci Lett. 2006;410:903.
73. Jacob S, Rajabally YA. Current proposed mechanisms of action of intravenous immunoglobulins in inflammatory neuropathies. Curr Neuro
pharmacol. 2009;7:33742.
74. Durandy A, Kaveri SV, Kuijpers TW, Basta M, Miescher S, Ravetch JV,
et al. Intravenous immunoglobulinsunderstanding properties and
mechanisms. Clin Exp Immunol. 2009;158 Suppl 1:213.
75. Kivity S, Katz U, Daniel N, Nussinovitch U, Papageorgiou N, Shoenfeld Y.
Evidence for the use of intravenous immunoglobulinsa review of the
literature. Clin Rev Allergy Immunol. 2010;38:20169.
76. Hughes RA, Dalakas MC, Cornblath DR, Latov N, Weksler ME,
Relkin N. Clinical applications of intravenous immunoglobulins in
neurology. Clin Exp Immunol. 2009;158 Suppl 1:3442.
77. Fillit H, Hess G, Hill J, Bonnet P, Toso C. IV immunoglobulin is associated
with a reduced risk of Alzheimer disease and related disorders. Neurology.
2009;73:1805.
78. Dodel R, Neff F, Noelker C, Pul R, Du Y, Bacher M, et al. Intravenous
immunoglobulins as a treatment for Alzheimers disease: rationale and
current evidence. Drugs. 2010;70:51328.
79. Neff F, Wei X, Nolker C, Bacher M, Du Y, Dodel R. Immunotherapy
and naturally occurring autoantibodies in neurodegenerative disorders.
Autoimmun Rev. 2008;7:5017.
80. Du Y, Wei X, Dodel R, Sommer N, Hampel H, Gao F, et al. Human
anti-beta-amyloid antibodies block beta-amyloid fibril formation and
prevent beta-amyloid-induced neurotoxicity. Brain. 2003;126:19359.
81. Patrias LM, Klaver AC, Coffey MP, Loeffler DA. Specific antibodies to
soluble alpha-synuclein conformations in intravenous immunoglobulin
preparations. Clin Exp Immunol. 2010;161:52735.
82. Szabo P, Relkin N, Weksler ME. Natural human antibodies to amyloid beta
peptide. Autoimmun Rev. 2008;7:41520.
83. Klaver AC, Finke JM, Digambaranath J, Balasubramaniam M, Loeffler DA.
Antibody concentrations to Abeta142monomer and soluble oligomers in
untreated and antibody-antigen-dissociated intravenous immunoglobulin
preparations. Int Immunopharmacol. 2010;10:1159.
84. Klaver AC, Patrias LM, Coffey MP, Finke JM, Loeffler DA. Measurement
of anti-Abeta142 antibodies in intravenous immunoglobulin with indirect
ELISA: the problem of nonspecific binding. J Neurosci Methods. 2010;
187:2639.
85. Szabo P, Mujalli DM, Rotondi ML, Sharma R, Weber A, Schwarz HP,
et al. Measurement of anti-beta amyloid antibodies in human blood.
J Neuroimmunol. 2010;227:16774.
86. Gomez-Isla T, Hollister R, West H, Mui S, Growdon JH, Petersen RC,
et al. Neuronal loss correlates with but exceeds neurofibrillary tangles in
Alzheimers disease. Ann Neurol. 1997;41:1724.
87. Van de Nes JA, Nafe R, Schlote W. Non-tau based neuronal degeneration
in Alzheimers diseasean immunocytochemical and quantitative study
in the supragranular layers of the middle temporal neocortex. Brain Res.
2008;1213:15265.

Journal of Central Nervous System Disease 2011:3

88. Kayed R, Pensalfini A, Margol L, Sokolov Y, Sarsoza F, Head E, et al.


Annular protofibrils are a structurally and functionally distinct type of
amyloid oligomer. J Biol Chem. 2009;284:42307.
89. Marx J. Alzheimers disease. A new take on tau. Science. 2007;316:14167.
90. Meraz-Rios MA, Lira-De Leon KI, Campos-Pena V, de AndaHernandez MA, Mena-Lopez R. Tau oligomers and aggregation in
Alzheimers d isease. J Neurochem. 2010;112:135367.
91. Lasagna-Reeves CA, Roi B, Guerrero-Muoz MJ, Castillo-Carranza DL,
Troncos JC, Kayed R, etal. Role of Tau oligomers in Parkinsons disease and
dementia with Lewy bodies. Movement Disorders. 2010;25:S181565.
92. Dance A, Landhuis E, Strobel G. The Society for Neuroscience
2009meeting report, part 2. J Alzheimers Dis. 2010;19:140915.
93. Andorfer C, Acker CM, Kress Y, Hof PR, Duff K, Davies P. Cell-cycle
reentry and cell death in transgenic mice expressing nonmutant human tau
isoforms. J Neurosci. 2005;25:544654.
94. Yoshiyama Y, Higuchi M, Zhang B, Huang SM, Iwata N, Saido TC,
etal. Synapse loss and microglial activation precede tangles in a P301S
tauopathy mouse model. Neuron. 2007;53:33751.
95. Berger Z, Roder H, Hanna A, Carlson A, Rangachari V, Yue M, et al.
Accumulation of pathological tau species and memory loss in a conditional
model of tauopathy. J Neurosci. 2007;27:365062.
96. Spires TL, Orne JD, SantaCruz K, Pitstick R, Carlson GA, Ashe KH, etal.
Region-specific dissociation of neuronal loss and neurofibrillary pathology
in a mouse model of tauopathy. Am J Pathol. 2006;168:1598607.
97. Maeda S, Sahara N, Saito Y, Murayama M, Yoshiike Y, Kim H, et al.
Granular tau oligomers as intermediates of tau filaments. Biochemistry.
2007;46:385661.
98. Maeda S, Sahara N, Saito Y, Murayama S, Ikai A, Takashima A. Increased
levels of granular tau oligomers: an early sign of brain aging and
Alzheimers disease. Neurosci Res. 2006;54:197201.
99. Duhem C, Dicato MA, Ries F. Side-effects of intravenous immune
globulins. Clin Exp Immunol. 1994;97 Suppl 1:7983.
100. Carbone J. Adverse reactions and pathogen safety of intravenous immunoglobulin. Curr Drug Saf. 2007;2:918.

Publish with Libertas Academica and


every scientist working in your field can
read your article
I would like to say that this is the most author-friendly
editing process I have experienced in over 150
publications. Thank you most sincerely.
The communication between your staff and me has
been terrific. Whenever progress is made with the
manuscript, I receive notice. Quite honestly, Ive
never had such complete communication with a
journal.
LA is different, and hopefully represents a kind of
scientific publication machinery that removes the
hurdles from free flow of scientific thought.

Your paper will be:



Available to your entire community
free of charge

Fairly and quickly peer reviewed

Yours! You retain copyright
http://www.la-press.com

73

Anda mungkin juga menyukai